1
|
Chong J, Chen Z, Ma J, He L, Zhu Y, Lu Z, Qiu Z, Chen C, Chen Y, Jiang F. Mechanistic investigation and the optimal dose based on baicalin in the treatment of ulcerative colitis-A preclinical systematic review and meta-analysis. BMC Gastroenterol 2025; 25:50. [PMID: 39901089 PMCID: PMC11792396 DOI: 10.1186/s12876-025-03629-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a type of inflammatory bowel disease, and current treatments often fall short, necessitating new therapeutic options. Baicalin shows therapeutic promise in UC animal models, but a systematic review is needed. METHODS A systematic search was conducted across databases including PubMed, EBSCO, Web of Science, and Science Direct, up to March 2024, identifying randomized controlled trials (RCTs) examining baicalin's impact on UC in animal models. Seventeen studies were selected through manual screening. Meta-analyses and subgroup analyses utilized Rev Man 5.3 and Stata 15.0 software to assess symptom improvement. RESULTS From 1304 citations, 17 were analyzed. Baicalin significantly modulated various biomarkers: HCS (SMD = -3.91), DAI (MD = -2.75), spleen index (MD = -12.76), MDA (SMD = -3.88), IL-6 (SMD = -10.59), IL-1β (SMD = -3.98), TNF-α (SMD = -8.05), NF-κB (SMD = -5.46), TLR4 (MD = -0.38), RORγ (MD = -0.89), MCP-1 (MD = -153.25), MPO (SMD = -7.34), Caspase-9 (MD = -0.93), Caspase-3 (MD = -0.45), FasL (MD = -1.20)) and enhanced BWC (MD = 0.06), CL (MD = 1.39), ZO-1 (MD = 0.44), SOD (SMD = 3.04), IL-10 mRNA (MD = 3.14), and FOXP3 (MD = 0.45) levels. Baicalin's actions may involve the PI3K/AKT, TLR4/NF-κB, IKK/IKB, Bcl-2/Bax, Th17/Treg, and TLRs/MyD88 pathways. Optimal therapeutic outcomes were predicted at dosages of 60-150 mg/kg over 10-14 weeks. CONCLUSION Baicalin demonstrates a multifaceted therapeutic potential in UC, attributed to its anti-inflammatory, antioxidant, anti-apoptotic, and intestinal barrier repair properties. While higher doses and longer treatments appear beneficial, further research, particularly human clinical trials, is necessary to verify its effectiveness and safety in people.
Collapse
Affiliation(s)
- Jinchen Chong
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zepeng Chen
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Jiaze Ma
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Linhai He
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Yijia Zhu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zhihua Lu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zhengxi Qiu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chen Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Yugen Chen
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, PR China.
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
- Jiangsu Collaborative Innovation Center of Chinese Medicine in Prevention and Treatment of Tumor, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
| | - Feng Jiang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, PR China.
| |
Collapse
|
2
|
Alexandru I, Davidescu L, Motofelea AC, Ciocarlie T, Motofelea N, Costachescu D, Marc MS, Suppini N, Șovrea AS, Coșeriu RL, Bondor DA, Bobeică LG, Crintea A. Emerging Nanomedicine Approaches in Targeted Lung Cancer Treatment. Int J Mol Sci 2024; 25:11235. [PMID: 39457017 PMCID: PMC11508987 DOI: 10.3390/ijms252011235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Lung cancer, the leading cause of cancer-related deaths worldwide, is characterized by its aggressive nature and poor prognosis. As traditional chemotherapy has the disadvantage of non-specificity, nanomedicine offers innovative approaches for targeted therapy, particularly through the development of nanoparticles that can deliver therapeutic agents directly to cancer cells, minimizing systemic toxicity and enhancing treatment efficacy. VEGF and VEGFR are shown to be responsible for activating different signaling cascades, which will ultimately enhance tumor development, angiogenesis, and metastasis. By inhibiting VEGF and VEGFR signaling pathways, these nanotherapeutics can effectively disrupt tumor angiogenesis and proliferation. This review highlights recent advancements in nanoparticle design, including lipid-based, polymeric, and inorganic nanoparticles, and their clinical implications in improving lung cancer outcomes, exploring the role of nanomedicine in lung cancer diagnoses and treatment.
Collapse
Affiliation(s)
- Isaic Alexandru
- Department X of General Surgery, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Lavinia Davidescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Tudor Ciocarlie
- Department VII Internal Medicine II, Discipline of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Nadica Motofelea
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
| | - Dan Costachescu
- Radiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Monica Steluta Marc
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Noemi Suppini
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Alina Simona Șovrea
- Department of Morphological Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Răzvan-Lucian Coșeriu
- Department of Microbiology, University of Medicine, Pharmacy, Science and Technology “George Emil Palade”, 540142 Târgu-Mures, Romania;
| | - Daniela-Andreea Bondor
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Laura-Gabriela Bobeică
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Andreea Crintea
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| |
Collapse
|
3
|
Chen Y, Tang Y, Li Y, Rui Y, Zhang P. Enhancing the Efficacy of Active Pharmaceutical Ingredients in Medicinal Plants through Nanoformulations: A Promising Field. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1598. [PMID: 39404324 PMCID: PMC11478102 DOI: 10.3390/nano14191598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
This article explores the emerging field of nanomedicine as a drug delivery system, aimed at enhancing the therapeutic efficacy of active pharmaceutical ingredients in medicinal plants. The traditional methods of applying medicinal plants present several limitations, such as low bioavailability, poor solubility, challenges in accurately controlling drug dosage, and inadequate targeting. Nanoformulations represent an innovative approach in drug preparation that employs nanotechnology to produce nanoscale particles or carriers, which are designed to overcome these limitations. Nanoformulations offer distinct advantages, significantly enhancing the solubility and bioavailability of drugs, particularly for the poorly soluble components of medicinal plants. These formulations effectively enhance solubility, thereby facilitating better absorption and utilization by the human body, which in turn improves drug efficacy. Furthermore, nanomedicine enables targeted drug delivery, ensuring precise administration to the lesion site and minimizing side effects on healthy tissues. Additionally, nanoformulations can regulate drug release rates, extend the duration of therapeutic action, and enhance the stability of treatment effects. However, nanoformulations present certain limitations and potential risks; their stability and safety require further investigation, particularly regarding the potential toxicity with long-term use. Nevertheless, nanomaterials demonstrate substantial potential in augmenting the efficacy of active pharmaceutical ingredients in medicinal plants, offering novel approaches and methodologies for their development and application.
Collapse
Affiliation(s)
- Yuhao Chen
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuying Tang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuanbo Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yukui Rui
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
- Tangshan Jinhai New Material Co., Ltd., Tangshan 063000, China
- Faculty of Resources and Environment, China Agricultural University, Shanghe County Baiqiao Town Science and Technology Courtyard, Jinan 250100, China
| | - Peng Zhang
- Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
4
|
Aslan C, Eraslan G. Effect of baicalin and baicalin-bovine serum albumin nanoparticle against bendiocarb exposure in rats. Toxicol Res (Camb) 2024; 13:tfae134. [PMID: 39233847 PMCID: PMC11369930 DOI: 10.1093/toxres/tfae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/17/2024] [Indexed: 09/06/2024] Open
Abstract
Background The aim of the study was to investigate the effect of baicalin and baicalin-bovine serum albumin nanoparticles against bendiocarb exposure in rats. Methods Eighty male Wistar Albino rats aged 4-6 weeks were used. Corn oil (vehicle) alone was administered to the control group. To other groups, BSA-nanoparticle equivalent to that binding baicalin at a dose of 20 mg/kg.bw, 20 mg/kg.bw baicalin, baicalin-BSA nanoparticle equivalent to that binding baicalin at a dose of 20 mg/kg.bw, 4 mg/kg.bw bendiocarb, combination of 4 mg/kg.bw bendiocarb and 20 mg/kg.bw baicalin, combination of 4 mg/kg.bw bendiocarb and BSA-nanoparticle equivalent to that binding baicalin at a dose of 20 mg/kg.bw and combination of 4 mg/kg.bw bendiocarb and baicalin-BSA nanoparticle equivalent to that binding baicalin at a dose of 20 mg/kg.bw was administered to animals by oral gavage with vehicle for 21 days, after which organs (liver, kidney, brain, testes, heart and lung) and blood samples were collected. Blood/tissue oxidative stress (MDA, NO, GSH, SOD, CAT, GSH-Px, GR, GST, G6PD), serum biochemical (glucose, triglyceride, cholesterol, BUN, creatinine, uric acid, total protein, albumin, LDH, AST, ALT, ALP and pseudocholinesterase) and liver and kidney apoptotic/anti-apoptotic (caspase 3, 9, p53, Bcl-2 and Bax) parameters were evaluated. Body weights/organ weights and plasma/liver bendiocarb analyses were obtained. Conclusion While bendiocarb administered alone caused oxidative stress/tissue damage, baicalin and baicalin-BSA nanoparticle showed a mitigating effect. However, this effect was more pronounced in the baicalin-BSA nanoparticle group. BSA-nanoparticle alone did not have a significant effect in reversing the adverse effect caused by bendiocarb.
Collapse
Affiliation(s)
- Coşkun Aslan
- Derinkuyu Emineana and Yaşar Ertaş Agriculture and Livestock Vocational School, Nevşehir Hacı Bektaş Veli University, Nevşehir, 50700, Turkey
| | - Gökhan Eraslan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, 38039, Turkey
| |
Collapse
|
5
|
Abdelrazek FN, Shalaby RA, Fahim SA, Essam RM, Anis SE, Attia YM, Abd El Malak NS. Novel fast dissolving freeze dried sublingual baicalin tablets for enhanced hepatoprotective effect: in-vitro characterization, cell viability, and in-vivo evaluation. Pharm Dev Technol 2024; 29:371-382. [PMID: 38613468 DOI: 10.1080/10837450.2024.2341243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/05/2024] [Indexed: 04/15/2024]
Abstract
Baicalin (BG), a natural product, has been used in the prevention and treatment of drug-induced liver injury (DILI); however, its poor solubility and extensive liver metabolism limit its pharmacological use. The aim of the present study was the formulation of fast-dissolving freeze-dried sublingual tablets (FFSTs) to increase BG dissolution, avoid first-pass metabolism, and overcome swallowing difficulties. FFSTs were prepared following a 23 factorial design. The effect of three independent variables namely matrix former, Maltodextrin, concentration (4%, and 6%), binder concentration (2%, and 3%), and binder type (Methocel E5, and Methocel E15) on the FFSTs' in-vitro disintegration time and percentage dissolution was studied along with other tablet characteristics. Differential scanning calorimetry, scanning electron microscopy, in-vitro HepG2 cell viability assay, and in-vivo characterization were also performed. F8 (6% Maltodextrin, 2% Mannitol, 2% Methocel E5), with desirability of 0.852, has been furtherly enhanced using 1%PEG (F10). F10 has achieved an in-vitro disintegration time of 41 secs, and 60.83% in-vitro dissolution after 2 min. Cell viability assay, in-vivo study in rats, and histopathological studies confirmed that pretreatment with F10 has achieved a significant hepatoprotective effect against acetaminophen-induced hepatotoxicity. The outcome of this study demonstrated that FFSTs may present a patient-friendly dosage form against DILI.
Collapse
Affiliation(s)
- Farida N Abdelrazek
- Pharmaceutics Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Rodayna A Shalaby
- Pharmaceutics Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Sally A Fahim
- Biochemistry Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Reham M Essam
- Biology department, School of Pharmacy, Newgiza University, Giza, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Shady E Anis
- Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yasmin M Attia
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Nevine S Abd El Malak
- Pharmaceutics Department, School of Pharmacy, Newgiza University, Giza, Egypt
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
6
|
Huang L, Huang XH, Yang X, Hu JQ, Zhu YZ, Yan PY, Xie Y. Novel nano-drug delivery system for natural products and their application. Pharmacol Res 2024; 201:107100. [PMID: 38341055 DOI: 10.1016/j.phrs.2024.107100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The development of natural products for potential new drugs faces obstacles such as unknown mechanisms, poor solubility, and limited bioavailability, which limit the broadened applicability of natural products. Therefore, there is a need for advanced pharmaceutical formulations of active compounds or natural products. In recent years, novel nano-drug delivery systems (NDDS) for natural products, including nanosuspensions, nanoliposomes, micelle, microemulsions/self-microemulsions, nanocapsules, and solid lipid nanoparticles, have been developed to improve solubility, bioavailability, and tissue distribution as well as for prolonged retention and enhanced permeation. Here, we updated the NDDS delivery systems used for natural products with the potential enhancement in therapeutic efficiency observed with nano-delivery systems.
Collapse
Affiliation(s)
- Li Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Xue-Hua Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Xi Yang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jia-Qin Hu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Yi-Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Pei-Yu Yan
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.
| | - Ying Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Sharawi ZW, Ibrahim IM, Abd-Alhameed EK, Althagafy HS, Jaber FA, Harakeh S, Hassanein EHM. Baicalin and lung diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1405-1419. [PMID: 37725153 DOI: 10.1007/s00210-023-02704-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
Studies focusing on natural products have been conducted worldwide, and the results suggest that their natural ingredients effectively treat a wide range of illnesses. Baicalin (BIA) is a glycoside derived from the flavonoid baicalein present in Scutellaria baicalensis of the Lamiaceae family. Interestingly, BIA has been shown to protect the lungs in several animal models used in numerous studies. Therefore, we fully analyzed the data of the studies that focused on BIA's lung protective function against various injuries and included them in this review. Interestingly, BIA exhibits promising effects against acute lung injury, lung fibrosis, pulmonary embolism, and lung remodelling associated with COPD, LPS, and paraquat insecticide. BAI exhibits anticancer activity against lung cancer. Additionally, BIA potently attenuates lung damage associated with infections. BIA primarily exerts its therapeutic effects by suppressing inflammation, oxidative stress immune response, and apoptosis pathways. Nrf2/HO-1, PI3K/Akt, NF-κB, STAT3, MAPKs, TLR4, and NLRP3 are important targets in the pulmonary therapeutic effects of BIA on different lung disease models. Consequently, we recommend using it in future potential clinical applications, its contribution to treatment guidelines, and translating its promising effects to clinical practice in lung diseases.
Collapse
Affiliation(s)
- Zeina W Sharawi
- Biological Sciences Department, Faculty of Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Fatima A Jaber
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah, 21589, Saudi Arabia
| | - Steve Harakeh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Yousef Abdul Lateef Jameel Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| |
Collapse
|
8
|
Sarma K, Akther MH, Ahmad I, Afzal O, Altamimi ASA, Alossaimi MA, Jaremko M, Emwas AH, Gautam P. Adjuvant Novel Nanocarrier-Based Targeted Therapy for Lung Cancer. Molecules 2024; 29:1076. [PMID: 38474590 DOI: 10.3390/molecules29051076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 03/14/2024] Open
Abstract
Lung cancer has the lowest survival rate due to its late-stage diagnosis, poor prognosis, and intra-tumoral heterogeneity. These factors decrease the effectiveness of treatment. They release chemokines and cytokines from the tumor microenvironment (TME). To improve the effectiveness of treatment, researchers emphasize personalized adjuvant therapies along with conventional ones. Targeted chemotherapeutic drug delivery systems and specific pathway-blocking agents using nanocarriers are a few of them. This study explored the nanocarrier roles and strategies to improve the treatment profile's effectiveness by striving for TME. A biofunctionalized nanocarrier stimulates biosystem interaction, cellular uptake, immune system escape, and vascular changes for penetration into the TME. Inorganic metal compounds scavenge reactive oxygen species (ROS) through their photothermal effect. Stroma, hypoxia, pH, and immunity-modulating agents conjugated or modified nanocarriers co-administered with pathway-blocking or condition-modulating agents can regulate extracellular matrix (ECM), Cancer-associated fibroblasts (CAF),Tyro3, Axl, and Mertk receptors (TAM) regulation, regulatory T-cell (Treg) inhibition, and myeloid-derived suppressor cells (MDSC) inhibition. Again, biomimetic conjugation or the surface modification of nanocarriers using ligands can enhance active targeting efficacy by bypassing the TME. A carrier system with biofunctionalized inorganic metal compounds and organic compound complex-loaded drugs is convenient for NSCLC-targeted therapy.
Collapse
Affiliation(s)
- Kangkan Sarma
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Md Habban Akther
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Manal A Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Preety Gautam
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| |
Collapse
|
9
|
Cai J, Hu Q, He Z, Chen X, Wang J, Yin X, Ma X, Zeng J. Scutellaria baicalensis Georgi and Their Natural Flavonoid Compounds in the Treatment of Ovarian Cancer: A Review. Molecules 2023; 28:5082. [PMID: 37446743 DOI: 10.3390/molecules28135082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common types of cancer in women with a high mortality rate, and the treatment of OC is prone to high recurrence rates and side effects. Scutellaria baicalensis (SB) is a herbal medicine with good anti-cancer activity, and several studies have shown that SB and its flavonoids have some anti-OC properties. This paper elucidated the common pathogenesis of OC, including cell proliferation and cell cycle regulation, cell invasion and metastasis, apoptosis and autophagy, drug resistance and angiogenesis. The mechanisms of SB and its flavonoids, wogonin, baicalein, baicalin, Oroxylin A, and scutellarein, in the treatment of OC, are revealed, such as wogonin inhibits proliferation, induces apoptosis, inhibits invasion and metastasis, and increases the cytotoxicity of the drug. Baicalein also inhibits vascular endothelial growth factor (VEGF) expression etc. Analyzing their advantages and disadvantages in treating OC provides a new perspective on the role of SB and its flavonoids in OC treatment. It serves as a resource for future OC research and development.
Collapse
Affiliation(s)
- Jiaying Cai
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhelin He
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiaoyan Chen
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Jian Wang
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiang Yin
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
10
|
Abeesh P, Guruvayoorappan C. Umbelliferone loaded PEGylated liposomes: preparation, characterization and its mitigatory effects on Dalton's ascites lymphoma development. 3 Biotech 2023; 13:216. [PMID: 37251730 PMCID: PMC10224889 DOI: 10.1007/s13205-023-03615-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Umbelliferone (UB) is a phenylpropanoid-based pharmacologically active agent with promising anti-tumor activities. However, complete elucidation of its therapeutic efficacy remains challenging due to low solubility and bioavailability. The present study aimed to develop a liposomal delivery system for UB to enhance its therapeutic efficacy against Dalton's ascites lymphoma tumor model. Umbelliferone-encapsulated nanoliposomes (nLUB) were prepared using the thin-film hydration method and performed a series of characterizations to confirm successful development. The nLUB showed a particle size of 116 ± 3.2 nm with a negative surface charge and encapsulation efficiency of 78%. In vitro study results showed that nLUB treatment significantly enhanced cellular uptake and apoptosis induction in lymphoma cells compared to free UB. nLUB treatment significantly stabilized body weight, reduced tumor growth, and improved the serum biochemical and hematological parameters of experimental animals, thereby improving their overall survivability compared to an free UB. Our result indicates that nanoencapsulation enhanced the therapeutic potential of UB, which may find its way to clinical application in the near future.
Collapse
Affiliation(s)
- Prathapan Abeesh
- Laboratory of Immunopharmacology and Experimental Therapeutics, Division of Cancer Research, Regional Cancer Centre, Medical College Campus, Thiruvananthapuram, Kerala 695 011 India
| | - Chandrasekharan Guruvayoorappan
- Laboratory of Immunopharmacology and Experimental Therapeutics, Division of Cancer Research, Regional Cancer Centre, Medical College Campus, Thiruvananthapuram, Kerala 695 011 India
| |
Collapse
|
11
|
Song S, Ding L, Liu G, Chen T, Zhao M, Li X, Li M, Qi H, Chen J, Wang Z, Wang Y, Ma J, Wang Q, Li X, Wang Z. The protective effects of baicalin for respiratory diseases: an update and future perspectives. Front Pharmacol 2023; 14:1129817. [PMID: 37007037 PMCID: PMC10060540 DOI: 10.3389/fphar.2023.1129817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/17/2023] Open
Abstract
Background: Respiratory diseases are common and frequent diseases. Due to the high pathogenicity and side effects of respiratory diseases, the discovery of new strategies for drug treatment is a hot area of research. Scutellaria baicalensis Georgi (SBG) has been used as a medicinal herb in China for over 2000 years. Baicalin (BA) is a flavonoid active ingredient extracted from SBG that BA has been found to exert various pharmacological effects against respiratory diseases. However, there is no comprehensive review of the mechanism of the effects of BA in treating respiratory diseases. This review aims to summarize the current pharmacokinetics of BA, baicalin-loaded nano-delivery system, and its molecular mechanisms and therapeutical effects for treating respiratory diseases.Method: This review reviewed databases such as PubMed, NCBI, and Web of Science from their inception to 13 December 2022, in which literature was related to “baicalin”, “Scutellaria baicalensis Georgi”, “COVID-19”, “acute lung injury”, “pulmonary arterial hypertension”, “asthma”, “chronic obstructive pulmonary disease”, “pulmonary fibrosis”, “lung cancer”, “pharmacokinetics”, “liposomes”, “nano-emulsions”, “micelles”, “phospholipid complexes”, “solid dispersions”, “inclusion complexes”, and other terms.Result: The pharmacokinetics of BA involves mainly gastrointestinal hydrolysis, the enteroglycoside cycle, multiple metabolic pathways, and excretion in bile and urine. Due to the poor bioavailability and solubility of BA, liposomes, nano-emulsions, micelles, phospholipid complexes, solid dispersions, and inclusion complexes of BA have been developed to improve its bioavailability, lung targeting, and solubility. BA exerts potent effects mainly by mediating upstream oxidative stress, inflammation, apoptosis, and immune response pathways. It regulates are the NF-κB, PI3K/AKT, TGF-β/Smad, Nrf2/HO-1, and ERK/GSK3β pathways.Conclusion: This review presents comprehensive information on BA about pharmacokinetics, baicalin-loaded nano-delivery system, and its therapeutic effects and potential pharmacological mechanisms in respiratory diseases. The available studies suggest that BA has excellent possible treatment of respiratory diseases and is worthy of further investigation and development.
Collapse
Affiliation(s)
- Siyu Song
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Lu Ding
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guangwen Liu
- GCP Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Tian Chen
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Meiru Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xueyan Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Min Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Qi
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinjin Chen
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ziyuan Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Ma
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Qi Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- *Correspondence: Xiangyan Li, ; Zeyu Wang,
| | - Zeyu Wang
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- *Correspondence: Xiangyan Li, ; Zeyu Wang,
| |
Collapse
|
12
|
Sharma A, Shambhwani D, Pandey S, Singh J, Lalhlenmawia H, Kumarasamy M, Singh SK, Chellappan DK, Gupta G, Prasher P, Dua K, Kumar D. Advances in Lung Cancer Treatment Using Nanomedicines. ACS OMEGA 2023; 8:10-41. [PMID: 36643475 PMCID: PMC9835549 DOI: 10.1021/acsomega.2c04078] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/13/2022] [Indexed: 06/01/2023]
Abstract
Carcinoma of the lungs is among the most menacing forms of malignancy and has a poor prognosis, with a low overall survival rate due to delayed detection and ineffectiveness of conventional therapy. Therefore, drug delivery strategies that may overcome undesired damage to healthy cells, boost therapeutic efficacy, and act as imaging tools are currently gaining much attention. Advances in material science have resulted in unique nanoscale-based theranostic agents, which provide renewed hope for patients suffering from lung cancer. Nanotechnology has vastly modified and upgraded the existing techniques, focusing primarily on increasing bioavailability and stability of anti-cancer drugs. Nanocarrier-based imaging systems as theranostic tools in the treatment of lung carcinoma have proven to possess considerable benefits, such as early detection and targeted therapeutic delivery for effectively treating lung cancer. Several variants of nano-drug delivery agents have been successfully studied for therapeutic applications, such as liposomes, dendrimers, polymeric nanoparticles, nanoemulsions, carbon nanotubes, gold nanoparticles, magnetic nanoparticles, solid lipid nanoparticles, hydrogels, and micelles. In this Review, we present a comprehensive outline on the various types of overexpressed receptors in lung cancer, as well as the various targeting approaches of nanoparticles.
Collapse
Affiliation(s)
- Akshansh Sharma
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| | | | - Sadanand Pandey
- Department
of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, Gyeongbuk 38541, South Korea
| | - Jay Singh
- Department
of Chemistry, Institute of Science, Banaras
Hindu University, Varanasi 221005, India
| | - Hauzel Lalhlenmawia
- Department
of Pharmacy, Regional Institute of Paramedical
and Nursing Sciences, Zemabawk, Aizawl, Mizoram 796017, India
| | - Murali Kumarasamy
- Department
of Biotechnology, National Institute of
Pharmaceutical Education and Research, Hajipur 844102, India
| | - Sachin Kumar Singh
- School
of Pharmaceutical Sciences, Lovely Professional
University, Phagwara 144411, India
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department
of Life Sciences, School of Pharmacy, International
Medical University, Kuala Lumpur 57000, Malaysia
| | - Gaurav Gupta
- Department
of Pharmacology, School of Pharmacy, Suresh
Gyan Vihar University, Jaipur 302017, India
- Department
of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical
and Technical Sciences, Saveetha University, Chennai 602117, India
- Uttaranchal
Institute of Pharmaceutical Sciences, Uttaranchal
University, Dehradun 248007, India
| | - Parteek Prasher
- Department
of Chemistry, University of Petroleum &
Energy Studies, Dehradun 248007, India
| | - Kamal Dua
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Discipline
of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Deepak Kumar
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| |
Collapse
|
13
|
Wang D, Li Y. Pharmacological effects of baicalin in lung diseases. Front Pharmacol 2023; 14:1188202. [PMID: 37168996 PMCID: PMC10164968 DOI: 10.3389/fphar.2023.1188202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
The flavonoids baicalin and baicalein were discovered in the root of Scutellaria baicalensis Georgi and are primarily used in traditional Chinese medicine, herbal supplements and healthcare. Recently, accumulated investigations have demonstrated the therapeutic benefits of baicalin in treating various lung diseases due to its antioxidant, anti-inflammatory, immunomodulatory, antiapoptotic, anticancer, and antiviral effects. In this review, the PubMed database and ClinicalTrials website were searched with the search string "baicalin" and "lung" for articles published between September 1970 and March 2023. We summarized the therapeutic role that baicalin plays in a variety of lung diseases, such as chronic obstructive pulmonary disease, asthma, pulmonary fibrosis, pulmonary hypertension, pulmonary infections, acute lung injury/acute respiratory distress syndrome, and lung cancer. We also discussed the underlying mechanisms of baicalin targeting in these lung diseases.
Collapse
Affiliation(s)
- Duoning Wang
- Chengdu Hi-tech Nanxili Jiuzheng Clinic, Chengdu, Sichuan, China
| | - Yi Li
- Chengdu Hi-tech Nanxili Jiuzheng Clinic, Chengdu, Sichuan, China
- *Correspondence: Yi Li, /
| |
Collapse
|
14
|
Gholami L, Ivari JR, Nasab NK, Oskuee RK, Sathyapalan T, Sahebkar A. Recent Advances in Lung Cancer Therapy Based on Nanomaterials: A Review. Curr Med Chem 2023; 30:335-355. [PMID: 34375182 DOI: 10.2174/0929867328666210810160901] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer is one of the commonest cancers with a significant mortality rate for both genders, particularly in men. Lung cancer is recognized as one of the leading causes of death worldwide, which threatens the lives of over 1.6 million people every day. Although cancer is the leading cause of death in industrialized countries, conventional anticancer medications are unlikely to increase patients' life expectancy and quality of life significantly. In recent years, there are significant advances in the development and applications of nanotechnology in cancer treatment. The superiority of nanostructured approaches is that they act more selectively than traditional agents. This progress led to the development of a novel field of cancer treatment known as nanomedicine. Various formulations based on nanocarriers, including lipids, polymers, liposomes, nanoparticles and dendrimers have opened new horizons in lung cancer therapy. The application and expansion of nano-agents lead to an exciting and challenging research era in pharmaceutical science, especially for the delivery of emerging anti-cancer agents. The objective of this review is to discuss the recent advances in three types of nanoparticle formulations for lung cancer treatments modalities, including liposomes, polymeric micelles, and dendrimers for efficient drug delivery. Afterward, we have summarized the promising clinical data on nanomaterials based therapeutic approaches in ongoing clinical studies.
Collapse
Affiliation(s)
- Leila Gholami
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Rouhani Ivari
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khandan Nasab
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom of Great Britain and Northern Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Wang L, Feng T, Su Z, Pi C, Wei Y, Zhao L. Latest research progress on anticancer effect of baicalin and its aglycone baicalein. Arch Pharm Res 2022; 45:535-557. [DOI: 10.1007/s12272-022-01397-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 07/11/2022] [Indexed: 11/02/2022]
|
16
|
Li L, Yu J, Chen Z, Zhang J. Improved Primary Lung Carcinoma Therapeutics Utilizing a Non-Invasive Approach of Combinatorial Drug Loaded Aerosolized Dry Inhaler Powder. J CLUST SCI 2022. [DOI: 10.1007/s10876-021-02103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
17
|
Xu W, Niu Y, Ai X, Xia C, Geng P, Zhu H, Zhou W, Huang H, Shi X. Liver-Targeted Nanoparticles Facilitate the Bioavailability and Anti-HBV Efficacy of Baicalin In Vitro and In Vivo. Biomedicines 2022; 10:biomedicines10040900. [PMID: 35453650 PMCID: PMC9025464 DOI: 10.3390/biomedicines10040900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 01/30/2023] Open
Abstract
The anti-hepatitis B virus (HBV) efficacy of baicalin (BA) is mediated by HBV-related hepatocyte nuclear factors (HNFs). However, this efficacy is severely limited by the low bioavailability of BA. Therefore, a novel liver-targeted BA liposome was constructed to promote the bioavailability and antiviral ability of BA. The results showed that apolipoprotein A1 (ApoA1)–modified liposomes (BAA1) significantly enhanced BA’s cellular uptake and specific distribution in the liver. Furthermore, the substantial inhibitory effects of BAA1 on HBsAg, HBeAg, HBV RNA, and HBV DNA were assessed in HB-infected cells and mice. Western blotting, co-immunoprecipitation, and transcriptomics analysis further revealed that the enhanced anti-HBV efficacy of BAA1 was attributed to the interaction between hepatocyte nuclear factors (HNFs) and estrogen receptors (ERs). Based on the findings, we propose that the ApoA1-modified liposomes aid BA in inhibiting HBV transcription and replication by augmenting its bioavailability and the HNFs–ERs axis.
Collapse
Affiliation(s)
- Weiming Xu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Yijun Niu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Xin Ai
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Chengjie Xia
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Ping Geng
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Haiyan Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Wei Zhou
- Department of Chemistry, Fudan University, 220 Han Dan Road, Shanghai 200433, China;
| | - Hai Huang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
| | - Xunlong Shi
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China; (W.X.); (Y.N.); (X.A.); (C.X.); (P.G.); (H.Z.); (H.H.)
- Correspondence: ; Tel.: +86-21-54237431; Fax: +86-21-51980037
| |
Collapse
|
18
|
Abeesh P, Guruvayoorappan C. Preparation and characterization of beta sitosterol encapsulated nanoliposomal formulation for improved delivery to cancer cells and evaluation of its anti-tumor activities against Daltons Lymphoma Ascites tumor models. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Kunimatsu R, Kimura A, Sakata S, Tsuka Y, Yoshimi Y, Abe T, Kado I, Yashima Y, Izumino J, Nakatani A, Kitagawa M, Miyauchi M, Takata T, Tanimoto K. Effects of baicalin on the proliferation and expression of OPG and RANKL in human cementoblast-lineage cells. J Dent Sci 2022; 17:162-169. [PMID: 35028034 PMCID: PMC8739232 DOI: 10.1016/j.jds.2021.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Background/purpose Baicalin, a natural bioactive flavonoid extracted from Scutellaria baicalensis Georgi, mediates bone metabolism, and recent studies have revealed that it has cell signaling properties. However, its biological functions in cementoblasts still remain unclear. This study therefore aimed to investigate the effects of baicalin on bone resorption markers, including osteoprotegerin (OPG) and receptor activator of nuclear factor-κβ ligand (RANKL), in human cementoblast-lineage cells, as well as their proliferation ability. Materials and methods Human cementoblast cell line (HCEM) cells were cultured and treated with 0, 0.01, 0.1, or 1 μM of baicalin. The proliferative capacity of cultured HCEM cells was analyzed using bromodeoxyuridine immunoassay and cell counting. The baicalin effect on OPG and RANKL expression was determined using quantitative polymerase chain reaction (qPCR) and western blotting. Furthermore, OPG expression was measured in 1 μM baicalin-treated HCEM cells in the presence or absence of the Wnt signaling pathway inhibitor, Dickkopf (Dkk)-1, using qPCR and western blotting. Results The addition of 0.01, 0.1, and 1 μM of baicalin did not significantly change the proliferative capacity of cultured HCEM cells. Compared with the non-supplemented group, baicalin increased and suppressed OPG and RANKL gene and protein expression, respectively, in a concentration-dependent manner. OPG mRNA and protein expression levels were increased by 1 μM baicalin, which was suppressed by Dkk-1 addition. Conclusion Baicalin enhanced OPG expression in HCEM cells through the Wnt/beta-catenin signaling pathway, which could contribute to periodontal tissue regeneration.
Collapse
Affiliation(s)
- Ryo Kunimatsu
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Aya Kimura
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuzo Sakata
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuji Tsuka
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Yoshimi
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takaharu Abe
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Isamu Kado
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuka Yashima
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Jin Izumino
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ayaka Nakatani
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masae Kitagawa
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan.,Tokuyama University, Tokuyama, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Development Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
20
|
Kong N, Chen X, Feng J, Duan T, Liu S, Sun X, Chen P, Pan T, Yan L, Jin T, Xiang Y, Gao Q, Wen C, Ma W, Liu W, Zhang M, Yang Z, Wang W, Zhang R, Chen B, Xie T, Sui X, Tao W. Baicalin induces ferroptosis in bladder cancer cells by downregulating FTH1. Acta Pharm Sin B 2021; 11:4045-4054. [PMID: 35024325 PMCID: PMC8727776 DOI: 10.1016/j.apsb.2021.03.036] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis is a non-apoptotic regulated cell death caused by iron accumulation and subsequent lipid peroxidation. Currently, the therapeutic role of ferroptosis on cancer is gaining increasing interest. Baicalin an active component in Scutellaria baicalensis Georgi with anticancer potential various cancer types; however, the effects of baicalein on bladder cancer and the underlying molecular mechanisms remain largely unknown. In the study, we investigated the effect of baicalin on bladder cancer cells 5637 and KU-19-19. As a result, we show baicalin exerted its anticancer activity by inducing apoptosis and cell death in bladder cancer cells. Subsequently, we for the first time demonstrate baicalin-induced ferroptotic cell death in vitro and in vivo, accompanied by reactive oxygen species (ROS) accumulation and intracellular chelate iron enrichment. The ferroptosis inhibitor deferoxamine but not necrostatin-1, chloroquine (CQ), N-acetyl-l-cysteine, l-glutathione reduced, or carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (Z-VAD-FMK) rescued baicalin-induced cell death, indicating ferroptosis contributed to baicalin-induced cell death. Mechanistically, we show that ferritin heavy chain 1 (FTH1) was a key determinant for baicalin-induced ferroptosis. Overexpression of FTH1 abrogated the anticancer effects of baicalin in both 5637 and KU19-19 cells. Taken together, our data for the first time suggest that the natural product baicalin exerts its anticancer activity by inducing FTH1-dependent ferroptosis, which will hopefully provide a prospective compound for bladder cancer treatment.
Collapse
Affiliation(s)
- Na Kong
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaying Chen
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiao Feng
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ting Duan
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Shuiping Liu
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Xueni Sun
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Peng Chen
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ting Pan
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Lili Yan
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ting Jin
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Yu Xiang
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Quan Gao
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Chengyong Wen
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Weirui Ma
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Wencheng Liu
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Mingming Zhang
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zuyi Yang
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Wengang Wang
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ruonan Zhang
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Bi Chen
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Tian Xie
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Xinbing Sui
- College of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
21
|
Bao J, Wu Z, Ishfaq M, Wang J, Miao Y, Niu D, Li R, Li J, Chen C. Pharmacokinetic/pharmacodynamic profiles of baicalin against Mycoplasma gallisepticum in an in vivo infection model. Poult Sci 2021; 100:101437. [PMID: 34547622 PMCID: PMC8463782 DOI: 10.1016/j.psj.2021.101437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 11/25/2022] Open
Abstract
Mycoplasma gallisepticum (M. gallisepticum), a devastating avian pathogen that commonly causes chronic respiratory disease in chicken, is responsible for tremendous economic losses to the poultry industry. Baicalin is the main constituent of Scutellaria baicalensis that shows potential therapeutic effects against M. gallisepticum. However, the pharmacokinetic/pharmacodynamics (PK/PD) profiles of baicalin against M. gallisepticum are not well understood. The main objective of the present study was to determine the relationship between the PK/PD index and efficacy of baicalin in the M. gallisepticum infection model in chickens. The experiments were carried out on 10-day-old chickens that were challenged with M. gallisepticum in the bilateral air sacs. While, baicalin was orally administrated once in a day for 3 consecutive days, started from d 3 postinfection. Ultra-performance liquid chromatography (UPLC) was used to evaluate the PK parameters of baicalin at doses of 200, 400, and 600 mg/kg in M. gallisepticum-infected chickens. Real-time PCR (RT-PCR) was used for the quantitative detection of M. gallisepticum in lungs. The PK and PD data were fitted to WinNonlin software to evaluate the PK/PD profiles of baicalin against M. gallisepticum. The minimum inhibitory concentration (MIC) of baicalin against M. gallisepticum strain Rlow was 31.25 µg/mL. The in vivo data suggested that baicalin concentration in the lung tissues was higher than plasma (1.21–1.73 times higher). The ratios of AUC24h/MIC of baicalin against bacteriostatic, bactericidal, and eradication were 0.62, 1.33, and 1.49 h, respectively. In conclusion, these results provided potential reference for future clinical dose selection of baicalin and evaluation of susceptibility breakpoints.
Collapse
Affiliation(s)
- Jiaxin Bao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Zhiyong Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Muhammad Ishfaq
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China; College of Computer Science, Huanggang Normal University, Huanggang, 438000, P. R. China
| | - Jian Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yusong Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Dong Niu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Rui Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, P. R. China..
| | - Chunli Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, P. R. China
| |
Collapse
|
22
|
Preparation and characterization of withaferin A loaded pegylated nanoliposomal formulation with high loading efficacy: In vitro and in vivo anti-tumour study. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112335. [PMID: 34474886 DOI: 10.1016/j.msec.2021.112335] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/08/2021] [Accepted: 07/22/2021] [Indexed: 12/09/2022]
Abstract
Withaferin A (WA) is a natural steroidal lactone with promising therapeutic applications. However, its clinical application is limited due to the low bioavailability and hydrophobic nature. In this study, we had prepared PEGylated nanoliposomal withaferin A (LWA) using thin-film hydration method. Dynamic light scattering, Transmission electron microscopy, and HPLC were used to investigate the impact of prepared formulations on the size, charge, morphology, and encapsulation efficiency of the LWA. The prepared nanoliposomal system had spherical vesicles, with the mean particle size of 125 nm and had an encapsulation efficiency of 83.65% with good stability. The characterization results indicated that nanoliposomal formulation is able to improve biocompatibility and bioavailability of WA. In vitro drug release study showed that LWA had an enhanced sustained drug release effect than the free drug. In vitro studies using ascites cell lines (DLA and EAC) showed that LWA treatment could induce apoptosis in ascites cells evidenced by acridine orange/ethidium bromide, Hoechst, and Giemsa staining. In vivo tumour study revealed that LWA treatment significantly reduced tumour growth and improved survival in DLA tumour bearing mice. In vivo results further demonstrated that LWA mitigated solid tumour development by regulating Ki-67 and cyclin D1 protein expression. The overall study results reveal that nanoliposome encapsulated WA exhibits therapeutic efficacy over WA in regulating tumour development as evidenced from ascites cell apoptosis as well as experimental tumour reduction studies.
Collapse
|
23
|
Sui X, Han X, Chen P, Wu Q, Feng J, Duan T, Chen X, Pan T, Yan L, Jin T, Xiang Y, Gao Q, Wen C, Ma W, Liu W, Zhang R, Chen B, Zhang M, Yang Z, Kong N, Xie T, Ding X. Baicalin Induces Apoptosis and Suppresses the Cell Cycle Progression of Lung Cancer Cells Through Downregulating Akt/mTOR Signaling Pathway. Front Mol Biosci 2021; 7:602282. [PMID: 33585556 PMCID: PMC7876332 DOI: 10.3389/fmolb.2020.602282] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/12/2020] [Indexed: 12/28/2022] Open
Abstract
Baicalin, as a natural active ingredient extracted and isolated from the traditional Chinese medicine Scutellaria baicalensis Georgi., has been potentially used in various areas for its antioxidative, antitumor, anti-inflammatory, and anti-proliferative activities. Although several studies have reported the antitumor effects of baicalin against various cancer types, its beneficial effects on lung cancer have not yet been elucidated. Therefore, the therapeutic effects and molecular mechanisms of baicalin on lung cancer cell lines H1299 and H1650 were investigated. Here, the results of its antitumor activity were shown. We found that Akt/mTOR pathway inhibition was the essential determinant in baicalin-induced cell cycle arrest. Furthermore, when the Akt Agonist SC79 or Akt plasmid transfection was performed, the antitumor effect of baicalin was significantly abrogated in both H1299 and H1650 cells. In conclusion, we found that baicalin exerted its antitumor activity mainly by inducing Akt-dependent cell cycle arrest and promoting apoptosis, which show great potential for developing a new drug for lung cancer treatment.
Collapse
Affiliation(s)
- Xinbing Sui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicine, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xuemeng Han
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Peng Chen
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jiao Feng
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Ting Duan
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Xiaying Chen
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Ting Pan
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Lili Yan
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Ting Jin
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Yu Xiang
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Quan Gao
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Chengyong Wen
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Weirui Ma
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Wencheng Liu
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Ruonan Zhang
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicine, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Bi Chen
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicine, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Mingming Zhang
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Zuyi Yang
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Na Kong
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- Department of Medical Oncology, School of Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicine, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
24
|
Baicalin mediated regulation of key signaling pathways in cancer. Pharmacol Res 2020; 164:105387. [PMID: 33352232 DOI: 10.1016/j.phrs.2020.105387] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023]
Abstract
Baicalin has been widely investigated against different types of malignancies both at the cellular and molecular levels over the past few years. Due to its remarkable anti-proliferative potential in numerous cancer cell lines, it has created immense interest as a potential chemotherapeutic modality compared to other flavonoids. Thus, this review focuses on the recent accomplishments of baicalin and its limitations in cancer prevention and treatment. Further, combination studies and nanoformulations using baicalin to treat cancer along with the metabolism, bioavailability, toxicity, and pharmacokinetics have been discussed. The present review explains biological source, and anti-proliferative potential of baicalin against cancers including breast, colon, hepatic, leukemia, lung, and skin, as well as the relevant mechanism of action to modulate diverse signaling pathways including apoptosis, cell cycle, invasion, and migration, angiogenesis, and autophagy. The anticancer mechanism of baicalin in orthotropic and xenograft mice models have been deliberated. The combination studies of baicalin in novel therapies as chemotherapeutic adjuvants have also been summarized. The low bioavailability, fast metabolism, and poor solubility, and other significant factors that limit the clinical use of baicalin have been examined as a challenge. The improvement in the pharmacokinetics and pharmacodynamics of baicalin with newer approaches and the gaps are highlighted, which could establish baicalin as an effective and safe compound for cancer treatment as well as help to translate its potential from bench to bedside.
Collapse
|
25
|
Liang Q, Xiang H, Li X, Luo C, Ma X, Zhao W, Chen J, Tian Z, Li X, Song X. Development of Rifapentine-Loaded PLGA-Based Nanoparticles: In vitro Characterisation and in vivo Study in Mice. Int J Nanomedicine 2020; 15:7491-7507. [PMID: 33116484 PMCID: PMC7547843 DOI: 10.2147/ijn.s257758] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/26/2020] [Indexed: 12/25/2022] Open
Abstract
Background Tuberculosis (TB) is a leading cause of death amongst infectious diseases. The poor response to antitubercular agents necessitates the long-term use of high drug doses, resulting in low patient compliance, which is the main reason for chemotherapy failure and contributes to the development of multidrug-resistant TB. Patient non-compliance has been a major obstacle in the successful management of TB. The aim of this work was to develop and characterise rifapentine (RPT)-loaded PLGA-based nanoparticles (NPs) for reducing dosing frequency. Methods RPT-loaded PLGA and PLGA–PEG NPs were prepared using premix membrane homogenisation combined with solvent evaporation method. The resulting NPs were characterised in terms of physicochemical characteristics, toxicity, cellular uptake and antitubercular activity. NPs were further evaluated for pharmacokinetic and biodistribution studies in mice. Results The resulting NPs showed suitable and safe physicochemical characteristics and could be taken up by macrophages. RPT-loaded NPs were more effective against Mycobacterium tuberculosis than free RPT. In vivo studies revealed that NPs could improve pharmacokinetic parameters, particularly for RPT/PLGA–PEG NPs. Moreover, both formulations had no toxicity to the organs of mice and could reduce hepatotoxicity. Conclusion The application of PLGA-based NPs as sustained-release delivery vehicles for RPT could prolong drug release, modify pharmacokinetics, increase antitubercular activity and diminish toxicity, thereby allowing low dosage and frequency.
Collapse
Affiliation(s)
- Qiuzhen Liang
- Department of Orthopaedics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, People's Republic of China
| | - Haibin Xiang
- Department of Orthopaedics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, People's Republic of China
| | - Xinyu Li
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, People's Republic of China
| | - Chunxia Luo
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, People's Republic of China
| | - Xuehong Ma
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, People's Republic of China
| | - Wenhui Zhao
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, People's Republic of China
| | - Jiangtao Chen
- Department of Orthopaedics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, People's Republic of China
| | - Zheng Tian
- Department of Orthopaedics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, People's Republic of China
| | - Xinxia Li
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, People's Republic of China
| | - Xinghua Song
- Department of Orthopaedic, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province 510630, People's Republic of China.,Department of Orthopaedic, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong Province 528303, People's Republic of China
| |
Collapse
|
26
|
Baicalin encapsulating lipid-surfactant conjugate based nanomicelles: Preparation, characterization and anticancer activity. Chem Phys Lipids 2020; 233:104978. [PMID: 32991905 DOI: 10.1016/j.chemphyslip.2020.104978] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/14/2020] [Accepted: 09/18/2020] [Indexed: 01/17/2023]
Abstract
Lung cancer is one of the most common malignant tumors and emerged as one of the leading causes of cancer-related death worldwide. Surgical resection can be a curative treatment for early stage but the most of lung cancer patients are diagnosed at an advanced stage when the pulmonary tumor has been invaded beyond the respiratory system. Therefore, chemotherapy is suitable for curing metastasized tumor. Baicalin (BL) is a flavonoid which has been studied in the treatment of several types of cancer including lung cancer. However, its low solubility in water and non-specificity impede its practical utilization. Hence, we have reported a stearic acid and pluronic F68 conjugated nanomicelles (PF68-SA) system to improve therapeutic efficacy of BL. Solvent evaporation method was used to prepare the BL-loaded PF68-SA nanomicelles (BLNM). The designed BLNM were characterized for the particle size, surface charge, critical micelle concentration, colloidal stability, morphology, and total drug content. BLNM formulation showed improved toxicity of BL against A549 human lung cancer cells in cytotoxicity assay. Further, apoptosis study also depicted BLNM-induced cell death in A549 cells. Therefore, the synthesized fatty acid-modified polymeric nanomicellar system could be useful in overcoming the stability and low therapeutic efficacy issues of hydrophobic anticancer drugs like BL and delivering them to the cancer cells.
Collapse
|
27
|
Guo LM, Xu XM, Zhao D, Cai XG, Zhou B. Biosynthesis, characterization of PLGA coated folate-mediated multiple drug loaded copper oxide (CuO) nanoparticles and it's cytotoxicity on nasopharyngeal cancer cell lines. AMB Express 2020; 10:160. [PMID: 32880769 PMCID: PMC7471236 DOI: 10.1186/s13568-020-01096-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Cytotoxicity of CuO nanoparticles (NPs) are an impediment in utilizing them as an effective nanocarriers of chemotherapeutic drugs for targeted drug delivery in nasopharyngeal cancer. In our current study, we have designed a two-step synthesis and coating of CuO NPs with different concentrations of PLGA (polylactide-co-glycolide) to reduce the cytotoxicity. This was further conjugated with folic acid to enhance targeting to specific tissue. The multiple drugs loaded in the NPs were two potent anticancer drugs doxorubicin and docetaxel. A complete characterization studies including micrographic analysis, zeta potential measurements, polydispersity index, Fourier transform infrared spectroscopy (FTIR), encapsulation and loading efficiencies, stability and in vitro release studies were done. Cytoxicity studies were done with MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, acridine orange/ethidium bromide and DAPI (4, 6-diamidino-2-phenylindole, dihydrochloride) staining procedures. Impediametric studies were also carried out to reinforce the reduction in cytotoxicity. Finally the cellular uptake of the NPs was seen. It was evident from the results that the multiple drugs loaded CuO NPs formed with PLGA coating were uniform, non-agglomerated in size ranging from 180 to 195 nm. The FTIR revealed no major changes in drug peaks. Encapsulation and loading efficiencies showed sufficient amount of drug being loaded into the NPs. The drug loaded NPs showed no change in size or zeta potential even after a period of 30 days. The cytotoxicity studies revealed significant reduction in toxicity after coating the surface treated with PLGA as evident from the microscopic analysis of cells. Hence the current study may be prioritized and further in vivo/in vitro studies may be carried out.
Collapse
Affiliation(s)
- Long-Mei Guo
- Department of Otolaryngology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37 of Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Xue-Mei Xu
- Department of Otolaryngology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37 of Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Dong Zhao
- Department of Otolaryngology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37 of Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Xun-Gong Cai
- Department of Otolaryngology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37 of Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Bin Zhou
- Department of Otolaryngology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37 of Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
28
|
Cai W, Geng C, Jiang L, Sun J, Chen B, Zhou Y, Yang B, Lu H. Encapsulation of gemcitabine in RGD-modified nanoliposomes improves breast cancer inhibitory activity. Pharm Dev Technol 2020; 25:640-648. [PMID: 32028816 DOI: 10.1080/10837450.2020.1727920] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 10/25/2022]
Abstract
In this study, RGD coated GEM liposomes were prepared by the emulsification-solvent evaporation method. The in vitro and in vivo characterizations were done to evaluate the feasibility of application. The mean particle size of the prepared liposomes was found to be 165.6 ± 15.7 nm. The entrapment efficiency and drug loading of the formulation were 82.4% ± 7.2% and 10.1% ± 1.4%, respectively. The liposomes were negatively charged with a zeta potential of -25.8 mV. The surface morphology of RGD-GEM liposomes was spherical and smooth. After three months of storage at different conditions, lyophilized liposomes appeared to be stable since they showed no collapse or contraction. The Weibull model was the most appropriate kinetic model for RGD-GEM liposomes, showing that the release of GEM from the liposomes was in the manners of both dissolution and diffusion. In vivo, the additive cytotoxicity of RGD-GEM-LPs in our study was caused by the presence of RGD which is more effective in the treatment of breast cancer devoid of toxicity to normal cells. Liposomes could also significantly extend the role of GEM in vivo and showed higher bioavailability than solution.
Collapse
Affiliation(s)
- Wei Cai
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Chunyan Geng
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Lei Jiang
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Jingping Sun
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Bin Chen
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Ying Zhou
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Binfeng Yang
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Hailin Lu
- Department of Oncology, Suzhou Ninth People's Hospital, Suzhou, China
| |
Collapse
|
29
|
Baicalin suppresses the cell cycle progression and proliferation of prostate cancer cells through the CDK6/FOXM1 axis. Mol Cell Biochem 2020; 469:169-178. [DOI: 10.1007/s11010-020-03739-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
|
30
|
Maltosyl-β-cyclodextrin mediated SupramolecularHost-Guest inclusion complex used for enhancing baicalin antioxidant activity and bioavailability. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Kang YY, Song J, Kim JY, Jung H, Yeo WS, Lim Y, Mok H. Byakangelicin as a modulator for improved distribution and bioactivity of natural compounds and synthetic drugs in the brain. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152963. [PMID: 31128487 DOI: 10.1016/j.phymed.2019.152963] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/08/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The elucidation of the biological roles of individual active compounds in terms of their in vivo bio-distribution and bioactivity could provide crucial information to understand how natural compounds work together as treatments for diseases. PURPOSE We examined the functional roles of Byakangelicin (Byn) to improve the brain accumulation of active compounds, e.g., umbelliferone (Umb), curcumin (Cur), and doxorubicin (Dox), and consequently to enhance their biological activities. METHODS Active compounds were administered intravenously to mice, with or without Byn, after which organs were isolated and visualized for their ex vivo fluorescence imaging to determine the bio-distribution of each active compound in vivo. For the in vivo bioactivity, Cur, either with or without Byn, was administered to a lipopolysaccharide (LPS)-induced neuro-inflammation model for 5 days, and its anti-inflammatory effects were examined by ELISA using a brain homogenate and serum. RESULTS We successfully demonstrated that the levels of active compounds (Umb, Cur, and Dox) in the brain, lung, and pancreas were greatly elevated by the addition of Byn via direct ex vivo fluorescence monitoring. In addition, sufficient accumulation of the active compound, Cur, greatly reduced LPS-induced neuro-inflammation in vivo. CONCLUSION Byn could serve as a modulator to allow improved brain accumulation of diverse active compounds (Umb, Cur, and Dox) and enhanced therapeutic effects.
Collapse
Affiliation(s)
- Yoon Young Kang
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jihyeon Song
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jun Yeong Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Heesun Jung
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Woon-Seok Yeo
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yoongho Lim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
32
|
Ancuceanu R, Dinu M, Dinu-Pirvu C, Anuţa V, Negulescu V. Pharmacokinetics of B-Ring Unsubstituted Flavones. Pharmaceutics 2019; 11:E370. [PMID: 31374885 PMCID: PMC6723510 DOI: 10.3390/pharmaceutics11080370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
B-ring unsubstituted flavones (of which the most widely known are chrysin, baicalein, wogonin, and oroxylin A) are 2-phenylchromen-4-one molecules of which the B-ring is devoid of any hydroxy, methoxy, or other substituent. They may be found naturally in a number of herbal products used for therapeutic purposes, and several have been designed by researchers and obtained in the laboratory. They have generated interest in the scientific community for their potential use in a variety of pathologies, and understanding their pharmacokinetics is important for a grasp of their optimal use. Based on a comprehensive survey of the relevant literature, this paper examines their absorption (with deglycosylation as a preliminary step) and their fate in the body, from metabolism to excretion. Differences among species (inter-individual) and within the same species (intra-individual) variability have been examined based on the available data, and finally, knowledge gaps and directions of future research are discussed.
Collapse
Affiliation(s)
- Robert Ancuceanu
- Department of Pharmaceutical Botany and Cell Biology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihaela Dinu
- Department of Pharmaceutical Botany and Cell Biology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.
| | - Cristina Dinu-Pirvu
- Department of Physical Chemistry and Colloidal Chemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest 020956, Romania
| | - Valentina Anuţa
- Department of Physical Chemistry and Colloidal Chemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest 020956, Romania
| | - Vlad Negulescu
- Department of Toxicology, Clinical Pharmacology and Psychopharmacology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
33
|
I El-Gogary R, Gaber SAA, Nasr M. Polymeric nanocapsular baicalin: Chemometric optimization, physicochemical characterization and mechanistic anticancer approaches on breast cancer cell lines. Sci Rep 2019; 9:11064. [PMID: 31363132 PMCID: PMC6667692 DOI: 10.1038/s41598-019-47586-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023] Open
Abstract
Baicalin is a multi-purpose flavonoid known for its anticancer properties, but its application is hindered by its low water solubility and bioavailability. Polymeric nanocapsules were proposed in this work as a promising system for enhancing baicalin delivery, and potentiating its anticancer properties. The characterization of nanocapsules was augmented with chemometric analysis, and the selected formulations were tested on two breast cancer cell lines (MCF-7 and MDA-MB-231), with mechanistic anticancer elucidation using MTT assay, confocal microscopy uptake, flow cytometry, mechanism of cell death, reactive oxygen species production, caspase 3/7 activity and death biomarker expression using quantitative real time PCR. Results showed that baicalin nanocapsules displayed favorable pharmaceutical properties; with the formulation variables affecting their properties elucidated using chemometric factorial analysis. Nanocapsules enhanced the anticancer activity of baicalin up to 216 times for MCF-7 cells and 31 times for MDA-MB-231 after 24 hr incubation. Cellular internalization of the fluorescently labeled nanocapsules was confirmed after 4 hr incubation for both cell lines. Apoptosis was the dominant cell death mechanism, with significant up-regulation of P53 in baicalin nanocapsules treated cells. Data here presented drive to further preclinical studies to investigate the delivery of baicalin polymeric nanocapsules and their anti-cancer activity.
Collapse
Affiliation(s)
- Riham I El-Gogary
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara A Abdel Gaber
- Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
34
|
Kulikov OA, Ageev VP, Marochkina EE, Dolgacheva IS, Minayeva OV, Inchina VI. Efficacy of liposomal dosage forms and hyperosmolar salines in experimental pharmacotherapy of acute lung injury. RESEARCH RESULTS IN PHARMACOLOGY 2019. [DOI: 10.3897/rrpharmacology.5.35529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction: Hypertonic sodium chloride solutions and liposomal drugs with pulmotropic effect are of great interest for the treatment of acute lung injury (ALI). The results of the studies on the efficacy of hypertonic solutions and liposomes in ALI treatment are currently controversial.Materials and methods: For the experiment, liposomes with dexamethasone, N-acetylcysteine (NAC), aprotinin and dye Cyanine-7 (Cy-7) were obtained. A liposome analysis was performed by means of spectrophotometry. ALI was modeled in rats by the administration of the damaging agents into the trachea. The experimental agents were injected once intravenously after the modeling of ALI. For experimental therapy used liposomal agents, 7.5% hypertonic saline (HS) and HyperHAES solutions in the respective groups. The efficacy of the therapy was assessed by the survival of animals, functional indicators of the cardiovascular and respiratory systems, and by the lung-body ratio. The biodistribution of liposomes after intravenous administration was investigated in mice through using a fluorescent dye Cy-7. The biodistribution of liposomes with Cy-7 was assessed using bioimaging according to the fluorescence intensity of internal organs (lungs, liver, and kidneys) and blood, expressed as dye concentration according to the calibration dependence of dye concentrarion on fluorescence intensity.Results and discussion: All the studied liposomal drugs were effective for the pharmacological correction of ALI. Hypertonic solutions, unlike liposomal drugs, were less likely to prevent the development of pulmonary edema. All the studied therapeutic agents increased the survival rate of the laboratory animals with ALI. The most effective experimental agent was liposomal dexamethasone. The use of drugs in form of simple liposomes with average diameter of 350 nm provided for a higher concentration of the drug in the lungs within the first 40 minutes after intravenous administration.Conclusion: Intravenous administration of liposomal forms is promising for the pharmacotherapy of acute lung injury.
Collapse
|
35
|
Li S, Li X, Ding J, Han L, Guo X. Anti-tumor efficacy of folate modified PLGA-based nanoparticles for the co-delivery of drugs in ovarian cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1271-1280. [PMID: 31114163 PMCID: PMC6497857 DOI: 10.2147/dddt.s195493] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/14/2019] [Indexed: 01/09/2023]
Abstract
Background: Ovarian cancer is a leading cause of death in gynecologic malignancies. The high mortality is mainly caused by advanced stage at presentation in most patients. Even after the combination of cytoreductive surgery and systemic platinum and taxane treatment, most patients relapse and eventually succumb to the disease. Therefore, there is an urgent need for new treatments. Purpose: A novel folate (FA)-targeted co-delivery of docetaxel (DTX) and gemcitabine (GEM) nanoparticles (NPs) was developed to overcome ovarian cancer. Materials and methods: Physicochemical characteristics of NPs such as size, morphology, and release profiles were explored. In vitro and in vivo studies were carried out to assess the efficacy of their antitumor activity in target cells. Results: FA modified DTX and GEM co-loaded NPs were prepared using the solvent evaporation method. The NPs with a particle size of ~120nm were stable in the observation period. The hemolysis results indicated that FA-PEG2000-PLGA was potentially feasible for targeted antitumor drug delivery through blood circulation. In vitro release study suggested that in comparison with the free drug, PLGA-DTX/GEM NPs and FA-PEG2000-PLGA-DTX/GEM NPs had sustained-release properties. However, there was no obvious difference between the two NPs with the same drug in the release profile. Ovarian cancer cells in vitro efficiently took up the non-targeted and FA-targeted NPs; improved cytotoxicity was observed in the FA-targeted NPs, showing a 3.59- fold drop in the IC50 in SKOV-3 cells as compared to DTX/GEM alone. Cellular uptake showed that through surface modification, more drugs entered the cell successfully. Pharmacodynamics results showed a statistically significant effect on the rate of reduction of tumor volume for FA-PEG2000-PLGA-DTX/GEM NPs than other groups and no toxicity of organs. Conclusion: The present study indicates that the FA-PEG2000-PLGA-DTX/GEM NPs provides a promising platform for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Shu Li
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiaofeng Li
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jianyi Ding
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Lingfei Han
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoqing Guo
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
36
|
Zheng Y, Shi S, Liu Y, Zhao Y, Sun Y. Targeted pharmacokinetics of polymeric micelles modified with glycyrrhetinic acid and hydrazone bond in H22 tumor-bearing mice. J Biomater Appl 2019; 34:141-151. [DOI: 10.1177/0885328219841487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yan Zheng
- College of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Shudan Shi
- College of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Yaru Liu
- College of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Yandan Zhao
- College of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Yuqi Sun
- College of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| |
Collapse
|
37
|
Lee SY, Koo JS, Yang M, Cho HJ. Application of temporary agglomeration of chitosan-coated nanoparticles for the treatment of lung metastasis of melanoma. J Colloid Interface Sci 2019; 544:266-275. [PMID: 30852352 DOI: 10.1016/j.jcis.2019.02.092] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/21/2022]
Abstract
Temporary association of chitosan (CS)-coated nanoparticles (NPs) including phloretin (Phl) in the blood stream can be applied to treat lung metastasis of melanoma. Phl was entrapped in poly(d,l-lactide-co-glycolide) (PLGA) NPs as an anticancer agent, whereas CS was decorated onto the outer surfaces of the Phl-loaded PLGA NPs (PLGA/Phl NPs). CS-coated PLGA/Phl NPs (CS-PLGA/Phl NPs) with mean hydrodynamic sizes of 342 nm, spherical shapes, unimodal size distribution, positive zeta potentials, and drug encapsulation efficiency larger than 90% were prepared. The presence of the CS layers in the outer surfaces of the CS-PLGA/Phl NPs was elucidated by X-ray photoelectron spectroscopy. Upon blending of the CS-PLGA/Phl NPs with serum albumin, microscale agglomerates formed and easily dissociated into individual NPs by applying external forces. A sustained Phl release from NPs and similar antiproliferation potential of the CS-PLGA/Phl NPs to that of Phl in melanoma (B16F10) cells were observed. After multiple dosing of developed NPs in mouse models with lung metastasis of melanoma, the CS-PLGA/Phl NPs group exhibited significantly lower lung weight and number of metastasis foci than the PLGA/Phl NPs group (p < 0.05). These results suggest that the transient transformation of NPs into microscale aggregates and their facile dissociation into individual NPs can be efficiently and safely applied for the treatment of lung metastasis of melanoma.
Collapse
Affiliation(s)
- Song Yi Lee
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ja Seong Koo
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Mingyu Yang
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyun-Jong Cho
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
38
|
Haider M, Hassan MA, Ahmed IS, Shamma R. Thermogelling Platform for Baicalin Delivery for Versatile Biomedical Applications. Mol Pharm 2018; 15:3478-3488. [PMID: 29953815 DOI: 10.1021/acs.molpharmaceut.8b00480] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Baicalin (BG) is a natural glycoside with several promising therapeutic and preventive applications. However, its pharmaceutical potential is compromised by its poor water solubility, complex oral absorption kinetics, and low bioavailability. In this work, BG was incorporated in a series of chitosan (Ch)/glycerophosphate (GP)-based thermosensitive hydrogel formulations to improve its water solubility and control its release profile. Molecular interactions between BG and GP were investigated using Fourier transform infrared spectroscopy (FT-IR), and the ability of GP to enhance the water solubility of BG was studied in different release media. Drug-loaded Ch/GP hydrogels were prepared and characterized for their gelation time, swelling ratio, and rheological properties in addition to surface and internal microstructure. Polyethylene glycol (PEG) 6000 and hydroxypropyl methyl cellulose (HPMC) were incorporated in the formulations at different ratios to study their effect on modulating the sol-gel behavior and the in vitro drug release. In vivo pharmacokinetic (PK) studies were carried out using a rabbit model to study the ability of drug-loaded Ch/GP thermosensitive hydrogels to control the absorption rate and improve the bioavailability of BG. Results showed that the solubility of BG was enhanced in the presence of GP, while the incorporation of PEG and/or HPMC had an impact on gelation time, rheological behavior, and rate of drug release in vitro. PK results obtained following buccal application of drug-loaded Ch/GP thermosensitive hydrogels to rabbits showed that the rate of BG absorption was controlled and the in vivo bioavailability was increased by 330% relative to BG aqueous oral suspension. The proposed Ch/GP thermosensitive hydrogel is an easily modifiable delivery platform that is not only capable of improving the solubility and bioavailability of BG following buccal administration but also can be suited for various local and injectable therapeutic applications.
Collapse
Affiliation(s)
- Mohamed Haider
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy , University of Sharjah , Sharjah 27272 , United Arab Emirates.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Cairo University , Cairo 11562 , Egypt
| | - Mariame A Hassan
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy , University of Sharjah , Sharjah 27272 , United Arab Emirates.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Cairo University , Cairo 11562 , Egypt
| | - Iman S Ahmed
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy , University of Sharjah , Sharjah 27272 , United Arab Emirates
| | - Rehab Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Cairo University , Cairo 11562 , Egypt
| |
Collapse
|
39
|
Hewei Z, Shuran M, Tian T, Ya X, Dandan Z, Tong W, Chunying H, Shujing Z. Inhibitory effect of Sumu (Lignum Sappan) plus Fuzi (Radix Aconiti Lateralis Praeparata) on a lung carcinoma model. J TRADIT CHIN MED 2018. [DOI: 10.1016/j.jtcm.2018.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Li S, Wang L, Li N, Liu Y, Su H. Combination lung cancer chemotherapy: Design of a pH-sensitive transferrin-PEG-Hz-lipid conjugate for the co-delivery of docetaxel and baicalin. Biomed Pharmacother 2017; 95:548-555. [DOI: 10.1016/j.biopha.2017.08.090] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/14/2017] [Accepted: 08/23/2017] [Indexed: 12/19/2022] Open
|
41
|
Santiago T, DeVaux RS, Kurzatkowska K, Espinal R, Herschkowitz JI, Hepel M. Surface-enhanced Raman scattering investigation of targeted delivery and controlled release of gemcitabine. Int J Nanomedicine 2017; 12:7763-7776. [PMID: 29123391 PMCID: PMC5661449 DOI: 10.2147/ijn.s149306] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Advanced and metastatic cancer forms are extremely difficult to treat and require high doses of chemotherapeutics, inadvertently affecting also healthy cells. As a result, the observed survival rates are very low. For instance, gemcitabine (GEM), one of the most effective chemotherapeutic drugs used for the treatment of breast and pancreatic cancers, sees only a 20% efficacy in penetrating cancer tissue, resulting in <5% survival rate in pancreatic cancer. Here, we present a method for delivering the drug that offers mitigation of side effects, as well as a targeted delivery and controlled release of the drug, improving its overall efficacy. By modifying the surface of gold nanoparticles (AuNPs) with covalently bonded thiol linkers, we have immobilized GEM on the nanoparticle (NP) through a pH-sensitive amide bond. This bond prevents the drug from being metabolized or acting on tissue at physiological pH 7.4, but breaks, releasing the drug at acidic pH, characteristic of cancer cells. Further functionalization of the NP with folic acid and/or transferrin (TF) offers a targeted delivery, as cancer cells overexpress folate and TF receptors, which can mediate the endocytosis of the NP carrying the drug. Thus, through the modification of AuNPs, we have been able to produce a nanocarrier containing GEM and folate/TF ligands, which is capable of targeted controlled-release delivery of the drug, reducing the side effects of the drug and increasing its efficacy. Here, we demonstrate the pH-dependent GEM release, using an ultrasensitive surface-enhanced Raman scattering spectroscopy to monitor the GEM loading onto the nanocarrier and follow its stimulated release. Further in vitro studies with model triple-negative breast cancer cell line MDA-MB-231 have corroborated the utility of the proposed nanocarrier method allowing the administration of high drug doses to targeted cancer cells.
Collapse
Affiliation(s)
- Ty Santiago
- Department of Chemistry, State University of New York at Potsdam, Potsdam
| | - Rebecca Sinnott DeVaux
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, USA
| | | | - Ricardo Espinal
- Department of Chemistry, State University of New York at Potsdam, Potsdam
| | - Jason I Herschkowitz
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Maria Hepel
- Department of Chemistry, State University of New York at Potsdam, Potsdam
| |
Collapse
|
42
|
Li C, Liu S, Luo G, Wang G, Zhang B, Nie Q. Comparison of plasma pharmacokinetics of Tanreqing solution between intratracheal aerosolization and intravenous injection in rats. Biomed Chromatogr 2017; 32. [PMID: 29027677 DOI: 10.1002/bmc.4116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/26/2017] [Accepted: 10/01/2017] [Indexed: 11/07/2022]
Abstract
A rapid ultra high performance liquid chromatography tandem mass spectrometry method was developed for the simultaneous analysis of baicalin, oroxylin A-7-O-β-d-glucoronide and chlorogenic acid in rats plasma, and applied to comparison of pharmacokinetics of Tanreqing solution between intratracheal aerosolization and intravenous injection. Results of the analytical method validation assay showed high sensitivity, accuracy and suitable recovery. Results of pharmacokinetics showed similar decline phases for baicalin, oroxylin A-7-O-β-d-glucoronide and chlorogenic acid in two different delivery routes. The half-lives of intratracheal aerosolization and intravenous injection were 0.90 and 1.22 h for baicalin, 0.47 and 0.17 h for oroxylin A-7-O-β-d-glucoronide and 0.22 and 0.13 h for chlorogenic acid, and this implies that compounds were retained in the lung for a relatively short time. This study was the first to provide important pharmacokinetics information for traditional Chinese medicine delivery to the lung.
Collapse
Affiliation(s)
- Cui Li
- China Academy of Chinese Medical Sciences, Beijing, China.,Institute of Chinese Material Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Siyu Liu
- The Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China
| | - Gan Luo
- Institute of Chinese Material Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guohua Wang
- Institute of Chinese Material Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoxian Zhang
- Institute of Chinese Material Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qixia Nie
- Institute of Chinese Material Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
43
|
Chen J, Wu Q, Luo L, Wang Y, Zhong Y, Dai HB, Sun D, Luo ML, Wu W, Wang GX. Dual tumor-targeted poly(lactic- co-glycolic acid)-polyethylene glycol-folic acid nanoparticles: a novel biodegradable nanocarrier for secure and efficient antitumor drug delivery. Int J Nanomedicine 2017; 12:5745-5760. [PMID: 28848351 PMCID: PMC5557624 DOI: 10.2147/ijn.s136488] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Further specific target-ability development of biodegradable nanocarriers is extremely important to promote their security and efficiency in antitumor drug-delivery applications. In this study, a facilely prepared poly(lactic-co-glycolic acid) (PLGA)-polyethylene glycol (PEG)-folic acid (FA) copolymer was able to self-assemble into nanoparticles with favorable hydrodynamic diameters of around 100 nm and negative surface charge in aqueous solution, which was expected to enhance intracellular antitumor drug delivery by advanced dual tumor-target effects, ie, enhanced permeability and retention induced the passive target, and FA mediated the positive target. Fluorescence-activated cell-sorting and confocal laser-scanning microscopy results confirmed that doxorubicin (model drug) loaded into PLGA-PEG-FA nanoparticles was able to be delivered efficiently into tumor cells and accumulated at nuclei. In addition, all hemolysis, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, and zebrafish-development experiments demonstrated that PLGA-PEG-FA nanoparticles were biocompatible and secure for biomedical applications, even at high polymer concentration (0.1 mg/mL), both in vitro and in vivo. Therefore, PLGA-PEG-FA nanoparticles provide a feasible controlled-release platform for secure and efficient antitumor drug delivery.
Collapse
Affiliation(s)
- Jia Chen
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
- Institute of Laboratory Animals, Sichuan Academy of Medical Science, Sichuan Provincial People’s Hospital, Chengdu
| | - Qi Wu
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| | - Li Luo
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| | - Han-Bin Dai
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| | - Da Sun
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
- Institute of Life Sciences, Wenzhou University, Wenzhou
| | - Mao-Ling Luo
- School of Medicine, Wuhan University, Wuhan, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| | - Gui-Xue Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing
| |
Collapse
|
44
|
High doses of baicalin induces kidney injury and fibrosis through regulating TGF-β/Smad signaling pathway. Toxicol Appl Pharmacol 2017; 333:1-9. [PMID: 28803990 DOI: 10.1016/j.taap.2017.08.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 08/09/2017] [Indexed: 11/22/2022]
Abstract
Baicalin is a major flavonoid compound purified from Scutellariae radix, which has been described as an herb in the Chinese Pharmacopoeia. Previous studies have suggested baicalin possessed extensive anti-inflammatory, anti-cancer, anti-viral properties. However, up to known, there have been no reports of safety and toxicity in the rats following oral administration of baicalin. In this present study, we showed the first evidence that treatment of baicalin (400, 800 and 1600mg/kg/day) induced significantly kidney injury and fibrosis. The collagen synthesis and fibrosis-related protein expression were increased in the kidney of Sprague-Dawley (SD) rats after treatment with high doses of baicalin. We further investigated the potential molecular mechanism of baicalin-mediated renal fibrosis and revealed that baicalin activated the transforming growth factor-β (TGF-β)/Smad signaling pathway in a dose-dependent manner. Moreover, we also observed that baicalin induced Smad3 interaction with transcriptional coactivator p300 accompanying with increment of Smad3 acetylation. Our results may contribute to better understanding of the future pharmacological and toxicological studies of Scutellaria baicalensis Georgi and its active compounds on the human disease.
Collapse
|
45
|
Ahammed V, Narayan R, Paul J, Nayak Y, Roy B, Shavi GV, Nayak UY. Development and in vivo evaluation of functionalized ritonavir proliposomes for lymphatic targeting. Life Sci 2017. [DOI: 10.1016/j.lfs.2017.06.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|